1
|
Tanaka R, Yamada K. Genomic and Reverse Translational Analysis Discloses a Role for Small GTPase RhoA Signaling in the Pathogenesis of Schizophrenia: Rho-Kinase as a Novel Drug Target. Int J Mol Sci 2023; 24:15623. [PMID: 37958606 PMCID: PMC10648424 DOI: 10.3390/ijms242115623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Schizophrenia is one of the most serious psychiatric disorders and is characterized by reductions in both brain volume and spine density in the frontal cortex. RhoA belongs to the RAS homolog (Rho) family and plays critical roles in neuronal development and structural plasticity via Rho-kinase. RhoA activity is regulated by GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). Several variants in GAPs and GEFs associated with RhoA have been reported to be significantly associated with schizophrenia. Moreover, several mouse models carrying schizophrenia-associated gene variants involved in RhoA/Rho-kinase signaling have been developed. In this review, we summarize clinical evidence showing that variants in genes regulating RhoA activity are associated with schizophrenia. In the last half of the review, we discuss preclinical evidence indicating that RhoA/Rho-kinase is a potential therapeutic target of schizophrenia. In particular, Rho-kinase inhibitors exhibit anti-psychotic-like effects not only in Arhgap10 S490P/NHEJ mice, but also in pharmacologic models of schizophrenia (methamphetamine- and MK-801-treated mice). Accordingly, we propose that Rho-kinase inhibitors may have antipsychotic effects and reduce cognitive deficits in schizophrenia despite the presence or absence of genetic variants in small GTPase signaling pathways.
Collapse
Affiliation(s)
- Rinako Tanaka
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan;
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan;
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Japan
| |
Collapse
|
2
|
Crystal Structure of the SH3 Domain of ASAP1 in Complex with the Proline Rich Motif (PRM) of MICAL1 Reveals a Unique SH3/PRM Interaction Mode. Int J Mol Sci 2023; 24:ijms24021414. [PMID: 36674928 PMCID: PMC9865144 DOI: 10.3390/ijms24021414] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
SH3 domains are common protein binding modules. The target sequence of SH3 domains is usually a proline-rich motif (PRM) containing a minimal "PxxP" sequence. The mechanism of how different SH3 domains specifically choose their targets from vast PxxP-containing sequences is still not very clear, as many reported SH3/PRM interactions are weak and promiscuous. Here, we identified the binding of the SH3 domain of ASAP1 to the PRM of MICAL1 with a sub-μM binding affinity, and determined the crystal structure of ASAP1-SH3 and MICAL1-PRM complex. Our structural and biochemical analyses revealed that the target-binding pocket of ASAP1-SH3 contains two negatively charged patches to recognize the "xPx + Px+" sequence in MICAL1-PRM and consequently strengthen the interaction, differing from the typical SH3/PRM interaction. This unique PRM-binding pocket is also found in the SH3 domains of GTPase Regulator associated with focal adhesion kinase (GRAF) and Src kinase associated phosphoprotein 1 (SKAP1), which we named SH3AGS. In addition, we searched the Swiss-Prot database and found ~130 proteins with the SH3AGS-binding PRM in silico. Finally, gene ontology analysis suggests that the strong interaction between the SH3AGS-containing proteins and their targets may play roles in actin cytoskeleton regulation and vesicle trafficking.
Collapse
|
3
|
Mallik B, Bhat S, Kumar V. Role of Bin‐Amphiphysin‐Rvs (BAR) domain proteins in mediating neuronal signaling and disease. Synapse 2022; 76:e22248. [DOI: 10.1002/syn.22248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Bhagaban Mallik
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Sajad Bhat
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Vimlesh Kumar
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| |
Collapse
|
4
|
Farooqi AA, Zahid R, Naureen H, Attar R, Gazouli M, Berardi R, Szelachowska J, Matkowski R, Pawlak E. Regulation of ROCK1/2 by long non-coding RNAs and circular RNAs in different cancer types. Oncol Lett 2022; 23:159. [PMID: 35399329 PMCID: PMC8987920 DOI: 10.3892/ol.2022.13279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 05/19/2021] [Indexed: 11/30/2022] Open
Abstract
Recent breakthroughs in high-throughput technologies have enabled the development of a better understanding of the functionalities of rho-associated protein kinases (ROCKs) under various physiological and pathological conditions. Since their discovery in the late 1990s, ROCKs have attracted the attention of interdisciplinary researchers due to their ability to pleiotropically modulate a myriad of cellular mechanisms. A rapidly growing number of published studies have started to shed light on the mechanisms underlying the regulation of ROCK1 and ROCK2 via long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) in different types of cancer. Detailed analyses have suggested that lncRNAs may be characteristically divided into oncogenic and tumor suppressor lncRNAs. Several exciting recent discoveries have also indicated how different lncRNAs and circRNAs modulate ROCK1/2 and mediate multistep cancer onset and progression. The present review chronicles the major advances that have been made in our understanding of the regulatory role of ROCK1/2 in different types of cancer, and how wide-ranging lncRNAs and circRNAs potentiate ROCK-driven signaling by blocking the targeting activities of tumor suppressor microRNAs.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering, Islamabad 54000, Pakistan
| | - Rabbia Zahid
- Institute of Chemistry, University of Punjab, Lahore 43000, Pakistan
| | - Humaira Naureen
- Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad 54000, Pakistan
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University 34280, Turkey
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens 54634, Greece
| | - Rossana Berardi
- Oncology Clinic-Marche Polytechnic University, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I-GM Lancisi-G Salesi di Ancona, I-60126 Ancona, Italy
| | - Jolanta Szelachowska
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
- Wroclaw Comprehensive Cancer Centre, 53-413 Wroclaw, Poland
| | - Rafał Matkowski
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
- Wroclaw Comprehensive Cancer Centre, 53-413 Wroclaw, Poland
| | - Edyta Pawlak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-013 Wroclaw, Poland
| |
Collapse
|
5
|
Wang K, Lu Y, Morrow DF, Xiao D, Xu C. Associations of ARHGAP26 Polymorphisms with Alzheimer's Disease and Cardiovascular Disease. J Mol Neurosci 2022; 72:1085-1097. [PMID: 35171450 DOI: 10.1007/s12031-022-01972-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
The Rho GTPase activating protein 26 (ARHGAP26) gene has been reported to be associated with neuropsychiatric diseases and neurodegenerative diseases including Parkinson's disease. We examined whether the ARHGAP26 gene is associated with Alzheimer's disease (AD) and/or cardiovascular disease (CVD). Multivariable logistic regression model was used to examine the associations of 154 single nucleotide polymorphisms (SNPs) within the ARHGAP26 gene with AD and CVD using the Alzheimer's Disease Neuroimaging Initiative 1 (ADNI-1) cohort. Fourteen SNPs were associated with AD (top SNP rs3776362 with p = 3.43 × 10-3), while 37 SNPs revealed associations with CVD (top SNP rs415235 with p = 2.06 × 10-4). Interestingly, 13 SNPs were associated with both AD and CVD. SNP rs3776362 was associated with CVD, Functional Activities Questionnaire (FAQ), and Clinical Dementia Rating Sum of Boxes (CDR-SB). A replication study using a Caribbean Hispanics sample showed that 17 SNPs revealed associations with AD, and 12 SNPs were associated with CVD. The third sample using a family-based study design showed that 9 SNPs were associated with AD, and 3 SNPs were associated with CVD. SNP rs6836509 within the ARHGAP10 gene (an important paralogon of ARHGAP26) was associated with AD and cerebrospinal fluid total tau (t-tau) level in the ADNI sample. Several SNPs were functionally important using the RegulomeDB, while a number of SNPs were associated with significant expression quantitative trait loci (eQTLs) using Genotype-Tissue Expression (GTEx) databases. In conclusion, genetic variants within ARHGAP26 were associated with AD and CVD. These findings add important new insights into the potentially shared pathogenesis of AD and CVD.
Collapse
Affiliation(s)
- Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Post Office Box 9600 - Office 6419, Morgantown, WV, 26506, USA.
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Deana F Morrow
- School of Social Work, West Virginia University, Morgantown, WV, 26506, USA
| | - Danqing Xiao
- Department of STEM, School of Arts and Sciences, Regis College, Weston, MA, 02493, USA
- McLean Imaging Center, McLean Hospital, MA, 02478, Belmont, USA
| | - Chun Xu
- Department of Health and Biomedical Sciences, College of Health Professions, University of Texas Rio Grande Valley, TX, 78520, Brownsville, USA.
| |
Collapse
|
6
|
The role of GTPase-activating protein ARHGAP26 in human cancers. Mol Cell Biochem 2021; 477:319-326. [PMID: 34716859 PMCID: PMC8755663 DOI: 10.1007/s11010-021-04274-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022]
Abstract
Rho GTPases are molecular switches that play an important role in regulating the behavior of a variety of tumor cells. RhoA GTPase-activating protein 26 (ARHGAP26) is a GTPase-activating protein and inhibits the activity of Rho GTPases by promoting the hydrolytic ability of Rho GTPases. It also affects tumorigenesis and progression of various tumors through several methods, including formation of abnormal fusion genes and circular RNA. This review summarizes the biological functions and molecular mechanisms of ARHGAP26 in different tumors, proposes the potential clinical value of ARHGAP26 in cancer treatment, and discusses current issues that need to be addressed.
Collapse
|
7
|
Lucken-Ardjomande Häsler S, Vallis Y, Pasche M, McMahon HT. GRAF2, WDR44, and MICAL1 mediate Rab8/10/11-dependent export of E-cadherin, MMP14, and CFTR ΔF508. J Cell Biol 2021; 219:151714. [PMID: 32344433 PMCID: PMC7199855 DOI: 10.1083/jcb.201811014] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 11/07/2019] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
In addition to the classical pathway of secretion, some transmembrane proteins reach the plasma membrane through alternative routes. Several proteins transit through endosomes and are exported in a Rab8-, Rab10-, and/or Rab11-dependent manner. GRAFs are membrane-binding proteins associated with tubules and vesicles. We found extensive colocalization of GRAF1b/2 with Rab8a/b and partial with Rab10. We identified MICAL1 and WDR44 as direct GRAF-binding partners. MICAL1 links GRAF1b/2 to Rab8a/b and Rab10, and WDR44 binds Rab11. Endogenous WDR44 labels a subset of tubular endosomes, which are closely aligned with the ER via binding to VAPA/B. With its BAR domain, GRAF2 can tubulate membranes, and in its absence WDR44 tubules are not observed. We show that GRAF2 and WDR44 are essential for the export of neosynthesized E-cadherin, MMP14, and CFTR ΔF508, three proteins whose exocytosis is sensitive to ER stress. Overexpression of dominant negative mutants of GRAF1/2, WDR44, and MICAL1 also interferes with it, facilitating future studies of Rab8/10/11-dependent exocytic pathways of central importance in biology.
Collapse
Affiliation(s)
| | - Yvonne Vallis
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Mathias Pasche
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Harvey T McMahon
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
8
|
The structure and function of protein kinase C-related kinases (PRKs). Biochem Soc Trans 2021; 49:217-235. [PMID: 33522581 PMCID: PMC7925014 DOI: 10.1042/bst20200466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 11/17/2022]
Abstract
The protein kinase C-related kinase (PRK) family of serine/threonine kinases, PRK1, PRK2 and PRK3, are effectors for the Rho family small G proteins. An array of studies have linked these kinases to multiple signalling pathways and physiological roles, but while PRK1 is relatively well-characterized, the entire PRK family remains understudied. Here, we provide a holistic overview of the structure and function of PRKs and describe the molecular events that govern activation and autoregulation of catalytic activity, including phosphorylation, protein interactions and lipid binding. We begin with a structural description of the regulatory and catalytic domains, which facilitates the understanding of their regulation in molecular detail. We then examine their diverse physiological roles in cytoskeletal reorganization, cell adhesion, chromatin remodelling, androgen receptor signalling, cell cycle regulation, the immune response, glucose metabolism and development, highlighting isoform redundancy but also isoform specificity. Finally, we consider the involvement of PRKs in pathologies, including cancer, heart disease and bacterial infections. The abundance of PRK-driven pathologies suggests that these enzymes will be good therapeutic targets and we briefly report some of the progress to date.
Collapse
|
9
|
Hada K, Wulaer B, Nagai T, Itoh N, Sawahata M, Sobue A, Mizoguchi H, Mori D, Kushima I, Nabeshima T, Ozaki N, Yamada K. Mice carrying a schizophrenia-associated mutation of the Arhgap10 gene are vulnerable to the effects of methamphetamine treatment on cognitive function: association with morphological abnormalities in striatal neurons. Mol Brain 2021; 14:21. [PMID: 33482876 PMCID: PMC7821731 DOI: 10.1186/s13041-021-00735-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/13/2021] [Indexed: 11/10/2022] Open
Abstract
We recently found a significant association between exonic copy-number variations in the Rho GTPase activating protein 10 (Arhgap10) gene and schizophrenia in Japanese patients. Special attention was paid to one patient carrying a missense variant (p.S490P) in exon 17, which overlapped with an exonic deletion in the other allele. Accordingly, we generated a mouse model (Arhgap10 S490P/NHEJ mice) carrying a missense variant and a coexisting frameshift mutation. We examined the spatiotemporal expression of Arhgap10 mRNA in the brain and found the highest expression levels in the cerebellum, striatum, and nucleus accumbens (NAc), followed by the frontal cortex in adolescent mice. The expression levels of phosphorylated myosin phosphatase-targeting subunit 1 and phosphorylated p21-activated kinases in the striatum and NAc were significantly increased in Arhgap10 S490P/NHEJ mice compared with wild-type littermates. Arhgap10 S490P/NHEJ mice exhibited a significant increase in neuronal complexity and spine density in the striatum and NAc. There was no difference in touchscreen-based visual discrimination learning between Arhgap10 S490P/NHEJ and wild-type mice, but a significant impairment of visual discrimination was evident in Arhgap10 S490P/NHEJ mice but not wild-type mice when they were treated with methamphetamine. The number of c-Fos-positive cells was significantly increased after methamphetamine treatment in the dorsomedial striatum and NAc core of Arhgap10 S490P/NHEJ mice. Taken together, these results suggested that schizophrenia-associated Arhgap10 gene mutations result in morphological abnormality of neurons in the striatum and NAc, which may be associated with vulnerability of cognition to methamphetamine treatment.
Collapse
Affiliation(s)
- Kazuhiro Hada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
| | - Bolati Wulaer
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192 Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
- Division of Behavioral Neuropharmacology, Project Office for Neuropsychological Research Center, Fujita Health University, Aichi, 470-1192 Japan
| | - Norimichi Itoh
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
| | - Akira Sobue
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560 Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560 Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, 466-8560 Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192 Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560 Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, 466-8560 Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8560 Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| |
Collapse
|
10
|
Dibus M, Brábek J, Rösel D. A Screen for PKN3 Substrates Reveals an Activating Phosphorylation of ARHGAP18. Int J Mol Sci 2020; 21:ijms21207769. [PMID: 33092266 PMCID: PMC7594087 DOI: 10.3390/ijms21207769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Protein kinase N3 (PKN3) is a serine/threonine kinase implicated in tumor progression of multiple cancer types, however, its substrates and effector proteins still remain largely understudied. In the present work we aimed to identify novel PKN3 substrates in a phosphoproteomic screen using analog sensitive PKN3. Among the identified putative substrates we selected ARHGAP18, a protein from RhoGAP family, for validation of the screen and further study. We confirmed that PKN3 can phosphorylate ARHGAP18 in vitro and we also characterized the interaction of the two proteins, which is mediated via the N-terminal part of ARHGAP18. We present strong evidence that PKN3-ARHGAP18 interaction is increased upon ARHGAP18 phosphorylation and that the phosphorylation of ARHGAP18 by PKN3 enhances its GAP domain activity and contributes to negative regulation of active RhoA. Taken together, we identified new set of potential PKN3 substrates and revealed a new negative feedback regulatory mechanism of Rho signaling mediated by PKN3-induced ARHGAP18 activation.
Collapse
Affiliation(s)
- Michal Dibus
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
- Correspondence:
| |
Collapse
|
11
|
Sekiguchi M, Sobue A, Kushima I, Wang C, Arioka Y, Kato H, Kodama A, Kubo H, Ito N, Sawahata M, Hada K, Ikeda R, Shinno M, Mizukoshi C, Tsujimura K, Yoshimi A, Ishizuka K, Takasaki Y, Kimura H, Xing J, Yu Y, Yamamoto M, Okada T, Shishido E, Inada T, Nakatochi M, Takano T, Kuroda K, Amano M, Aleksic B, Yamomoto T, Sakuma T, Aida T, Tanaka K, Hashimoto R, Arai M, Ikeda M, Iwata N, Shimamura T, Nagai T, Nabeshima T, Kaibuchi K, Yamada K, Mori D, Ozaki N. ARHGAP10, which encodes Rho GTPase-activating protein 10, is a novel gene for schizophrenia risk. Transl Psychiatry 2020; 10:247. [PMID: 32699248 PMCID: PMC7376022 DOI: 10.1038/s41398-020-00917-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/12/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia (SCZ) is known to be a heritable disorder; however, its multifactorial nature has significantly hampered attempts to establish its pathogenesis. Therefore, in this study, we performed genome-wide copy-number variation (CNV) analysis of 2940 patients with SCZ and 2402 control subjects and identified a statistically significant association between SCZ and exonic CNVs in the ARHGAP10 gene. ARHGAP10 encodes a member of the RhoGAP superfamily of proteins that is involved in small GTPase signaling. This signaling pathway is one of the SCZ-associated pathways and may contribute to neural development and function. However, the ARHGAP10 gene is often confused with ARHGAP21, thus, the significance of ARHGAP10 in the molecular pathology of SCZ, including the expression profile of the ARHGAP10 protein, remains poorly understood. To address this issue, we focused on one patient identified to have both an exonic deletion and a missense variant (p.S490P) in ARHGAP10. The missense variant was found to be located in the RhoGAP domain and was determined to be relevant to the association between ARHGAP10 and the active form of RhoA. We evaluated ARHGAP10 protein expression in the brains of reporter mice and generated a mouse model to mimic the patient case. The model exhibited abnormal emotional behaviors, along with reduced spine density in the medial prefrontal cortex (mPFC). In addition, primary cultured neurons prepared from the mouse model brain exhibited immature neurites in vitro. Furthermore, we established induced pluripotent stem cells (iPSCs) from this patient, and differentiated them into tyrosine hydroxylase (TH)-positive neurons in order to analyze their morphological phenotypes. TH-positive neurons differentiated from the patient-derived iPSCs exhibited severe defects in both neurite length and branch number; these defects were restored by the addition of the Rho-kinase inhibitor, Y-27632. Collectively, our findings suggest that rare ARHGAP10 variants may be genetically and biologically associated with SCZ and indicate that Rho signaling represents a promising drug discovery target for SCZ treatment.
Collapse
Affiliation(s)
- Mariko Sekiguchi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akira Sobue
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Itaru Kushima
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.437848.40000 0004 0569 8970Medical Genomics Center, Nagoya University Hospital, Nagoya, Aichi Japan
| | - Chenyao Wang
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yuko Arioka
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.437848.40000 0004 0569 8970Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Aichi Japan
| | - Hidekazu Kato
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akiko Kodama
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Hisako Kubo
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Norimichi Ito
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Masahito Sawahata
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kazuhiro Hada
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Ryosuke Ikeda
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Mio Shinno
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Chikara Mizukoshi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Keita Tsujimura
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akira Yoshimi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kanako Ishizuka
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yuto Takasaki
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Hiroki Kimura
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Jingrui Xing
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yanjie Yu
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Maeri Yamamoto
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Takashi Okada
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Emiko Shishido
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Toshiya Inada
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Masahiro Nakatochi
- grid.27476.300000 0001 0943 978XDivision of Data Science, Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Tetsuya Takano
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Keisuke Kuroda
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Mutsuki Amano
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Branko Aleksic
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Takashi Yamomoto
- grid.257022.00000 0000 8711 3200Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tetsushi Sakuma
- grid.257022.00000 0000 8711 3200Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tomomi Aida
- grid.265073.50000 0001 1014 9130Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohichi Tanaka
- grid.265073.50000 0001 1014 9130Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryota Hashimoto
- grid.419280.60000 0004 1763 8916Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan ,grid.136593.b0000 0004 0373 3971Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan ,grid.136593.b0000 0004 0373 3971Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Makoto Arai
- grid.272456.0Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masashi Ikeda
- grid.256115.40000 0004 1761 798XDepartment of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nakao Iwata
- grid.256115.40000 0004 1761 798XDepartment of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Teppei Shimamura
- grid.27476.300000 0001 0943 978XDivision of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Taku Nagai
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory Fujita Health University, Graduate School of Health Sciences & Aino University, Toyoake, Aichi Japan
| | - Kozo Kaibuchi
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan. .,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan. .,Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan.
| | - Norio Ozaki
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| |
Collapse
|
12
|
Ohta K, Matsumoto Y, Nishio M. Common and unique mechanisms of filamentous actin formation by viruses of the genus Orthorubulavirus. Arch Virol 2020; 165:799-807. [PMID: 32100137 DOI: 10.1007/s00705-020-04565-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/28/2020] [Indexed: 11/28/2022]
Abstract
We previously found that infection with human parainfluenza virus type 2 (hPIV-2), a member of the genus Orthorubulavirus, family Paramyxoviridae, causes filamentous actin (F-actin) formation to promote viral growth. In the present study, we investigated whether similar regulation of F-actin formation is observed in infections with other rubulaviruses, such as parainfluenza virus type 5 (PIV-5) and simian virus 41 (SV41). Infection with these viruses caused F-actin formation and RhoA activation, which promoted viral growth. These results indicate that RhoA-induced F-actin formation is important for efficient growth of these rubulaviruses. Only SV41 and hPIV-2 V and P proteins bound to Graf1, while the V and P proteins of PIV-5, mumps virus, and hPIV-4 did not bind to Graf1. In contrast, the V proteins of these rubulaviruses bound to both inactive RhoA and profilin 2. These results suggest that there are common and unique mechanisms involved in regulation of F-actin formation by members of the genus Orthorubulavirus.
Collapse
Affiliation(s)
- Keisuke Ohta
- Department of Microbiology, School of Medicine, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509, Japan
| | - Yusuke Matsumoto
- Department of Microbiology, School of Medicine, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509, Japan
| | - Machiko Nishio
- Department of Microbiology, School of Medicine, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
13
|
Li Y, Zeng B, Li Y, Zhang C, Ren G. Downregulated expression of ARHGAP10 correlates with advanced stage and high Ki-67 index in breast cancer. PeerJ 2019; 7:e7431. [PMID: 31396458 PMCID: PMC6679923 DOI: 10.7717/peerj.7431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/08/2019] [Indexed: 12/19/2022] Open
Abstract
Background Rho GTPase-activating protein 10 (ARHGAP10), which catalyzes the conversion of active Rho GTPase to the inactive form, is downregulated in some cancers. However, little is known about ARHGAP10 in breast cancer. Methods The transcriptional expression level of ARHGAP10 in breast cancer was analyzed with the data downloaded from The Cancer Genome Atlas (TCGA) and Oncomine, then verified by reverse-transcription quantitative polymerase chain reaction (RT-qPCR) in 30 pairs of breast cancer tissues and the corresponding adjacent normal tissues. ARHGAP10 protein expression was examined by immunohistochemistry (IHC) in 190 breast cancer and 30 corresponding adjacent normal breast tissue samples. The associations between ARHGAP10 expression and clinicopathological characteristics of patients were analyzed, and Kaplan-Meier Plotter was used to assess the relationship between ARHGAP10 and relapse-free survival (RFS). Different expression levels of ARHGAP10 in response to chemotherapy agents were determined by GEO2R online tool. The potential biological functions of ARHGAP10 were analyzed by Gene Set Enrichment Analysis (GSEA) using data downloaded from TCGA. Results ARHGAP10 mRNA and protein expression was lower in breast cancer tissues than in adjacent normal tissues. Low expression of ARHGAP10 was associated with advanced clinical TNM (cTNM) stage (p b = 0.001) and high Ki-67 index (p = 0.015). Low expression of ARHGAP10 indicated worse RFS (p = 0.0015) and a poor response to chemotherapy (p = 0.006). GSEA results showed that ARHGAP10 was involved in signaling pathways including protein export, nucleotide excision repair, base excision repair, focal adhesion, JAK-STAT pathway and the actin cytoskeleton.
Collapse
Affiliation(s)
- Yujing Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Beilei Zeng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14
|
Luo W, Janoštiak R, Tolde O, Ryzhova LM, Koudelková L, Dibus M, Brábek J, Hanks SK, Rosel D. ARHGAP42 is activated by Src-mediated tyrosine phosphorylation to promote cell motility. J Cell Sci 2017; 130:2382-2393. [PMID: 28584191 PMCID: PMC5536916 DOI: 10.1242/jcs.197434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
The tyrosine kinase Src acts as a key regulator of cell motility by phosphorylating multiple protein substrates that control cytoskeletal and adhesion dynamics. In an earlier phosphotyrosine proteomics study, we identified a novel Rho-GTPase activating protein, now known as ARHGAP42, as a likely biologically relevant Src substrate. ARHGAP42 is a member of a family of RhoGAPs distinguished by tandem BAR-PH domains lying N-terminal to the GAP domain. Like other family members, ARHGAP42 acts preferentially as a GAP for RhoA. We show that Src principally phosphorylates ARHGAP42 on tyrosine 376 (Tyr-376) in the short linker between the BAR-PH and GAP domains. The expression of ARHGAP42 variants in mammalian cells was used to elucidate its regulation. We found that the BAR domain is inhibitory toward the GAP activity of ARHGAP42, such that BAR domain deletion resulted in decreased active GTP-bound RhoA and increased cell motility. With the BAR domain intact, ARHGAP42 GAP activity could be activated by phosphorylation of Tyr-376 to promote motile cell behavior. Thus, phosphorylation of ARHGAP42 Tyr-376 is revealed as a novel regulatory event by which Src can affect actin dynamics through RhoA inhibition.
Collapse
Affiliation(s)
- Weifeng Luo
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
| | - Radoslav Janoštiak
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
| | - Ondřej Tolde
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy 25242, Czech Republic
| | - Larisa M Ryzhova
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lenka Koudelková
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy 25242, Czech Republic
| | - Michal Dibus
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy 25242, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy 25242, Czech Republic
| | - Steven K Hanks
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, 12843, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy 25242, Czech Republic
| |
Collapse
|
15
|
Graf1 Controls the Growth of Human Parainfluenza Virus Type 2 through Inactivation of RhoA Signaling. J Virol 2016; 90:9394-405. [PMID: 27512058 DOI: 10.1128/jvi.01471-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Rho GTPases are involved in a variety of cellular activities and are regulated by guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). We found that the activation of Rho GTPases by lysophosphatidic acid promotes the growth of human parainfluenza virus type 2 (hPIV-2). Furthermore, hPIV-2 infection causes activation of RhoA, a Rho GTPase. We hypothesized that Graf1 (also known as ARHGAP26), a GAP, regulates hPIV-2 growth by controlling RhoA signaling. Immunofluorescence analysis showed that hPIV-2 infection altered Graf1 localization from a homogenous distribution within the cytoplasm to granules. Graf1 colocalized with hPIV-2 P, NP, and L proteins. Graf1 interacts with P and V proteins via their N-terminal common region, and the C-terminal Src homology 3 domain-containing region of Graf1 is important for these interactions. In HEK293 cells constitutively expressing Graf1, hPIV-2 growth was inhibited, and RhoA activation was not observed during hPIV-2 infection. In contrast, Graf1 knockdown restored hPIV-2 growth and RhoA activation. Overexpression of hPIV-2 P and V proteins enhanced hPIV-2-induced RhoA activation. These results collectively suggested that hPIV-2 P and V proteins enhanced hPIV-2 growth by binding to Graf1 and that Graf1 inhibits hPIV-2 growth through RhoA inactivation. IMPORTANCE Robust growth of hPIV-2 requires Rho activation. hPIV-2 infection causes RhoA activation, which is suppressed by Graf1. Graf1 colocalizes with viral RNP (vRNP) in hPIV-2-infected cells. We found that Graf1 interacts with hPIV-2 P and V proteins. We also identified regions in these proteins which are important for this interaction. hPIV-2 P and V proteins enhanced the hPIV-2 growth via binding to Graf1, while Graf1 inhibited hPIV-2 growth through RhoA inactivation.
Collapse
|
16
|
Mukai H, Muramatsu A, Mashud R, Kubouchi K, Tsujimoto S, Hongu T, Kanaho Y, Tsubaki M, Nishida S, Shioi G, Danno S, Mehruba M, Satoh R, Sugiura R. PKN3 is the major regulator of angiogenesis and tumor metastasis in mice. Sci Rep 2016; 6:18979. [PMID: 26742562 PMCID: PMC4705536 DOI: 10.1038/srep18979] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/02/2015] [Indexed: 01/13/2023] Open
Abstract
PKN, a conserved family member related to PKC, was the first protein kinase identified as a target of the small GTPase Rho. PKN is involved in various functions including cytoskeletal arrangement and cell adhesion. Furthermore, the enrichment of PKN3 mRNA in some cancer cell lines as well as its requirement in malignant prostate cell growth suggested its involvement in oncogenesis. Despite intensive research efforts, physiological as well as pathological roles of PKN3 in vivo remain elusive. Here, we generated mice with a targeted deletion of PKN3. The PKN3 knockout (KO) mice are viable and develop normally. However, the absence of PKN3 had an impact on angiogenesis as evidenced by marked suppressions of micro-vessel sprouting in ex vivo aortic ring assay and in vivo corneal pocket assay. Furthermore, the PKN3 KO mice exhibited an impaired lung metastasis of melanoma cells when administered from the tail vein. Importantly, PKN3 knock-down by small interfering RNA (siRNA) induced a glycosylation defect of cell-surface glycoproteins, including ICAM-1, integrin β1 and integrin α5 in HUVECs. Our data provide the first in vivo genetic demonstration that PKN3 plays critical roles in angiogenesis and tumor metastasis, and that defective maturation of cell surface glycoproteins might underlie these phenotypes.
Collapse
Affiliation(s)
- Hideyuki Mukai
- Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - Aiko Muramatsu
- Graduate School of Science and Technology, Kobe University, Kobe 657-8501, Japan
| | - Rana Mashud
- Graduate School of Medicine, Kobe University, Kobe 657-8501, Japan
| | - Koji Kubouchi
- Laboratory of Molecular Pharmacogenomics, School of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Sho Tsujimoto
- Laboratory of Molecular Pharmacogenomics, School of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Tsunaki Hongu
- Graduate School of Comprehensive Human Sciences, Institute of Basic Medical Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yasunori Kanaho
- Graduate School of Comprehensive Human Sciences, Institute of Basic Medical Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kinki University School of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kinki University School of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan
| | - Go Shioi
- Genetic Engineering Team, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami,Chuou-ku, Kobe 650-0047
| | - Sally Danno
- Graduate School of Medicine, Kobe University, Kobe 657-8501, Japan
| | - Mona Mehruba
- Graduate School of Medicine, Kobe University, Kobe 657-8501, Japan
| | - Ryosuke Satoh
- Laboratory of Molecular Pharmacogenomics, School of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Reiko Sugiura
- Laboratory of Molecular Pharmacogenomics, School of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| |
Collapse
|
17
|
Jarius S, Wildemann B. 'Medusa head ataxia': the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 2: Anti-PKC-gamma, anti-GluR-delta2, anti-Ca/ARHGAP26 and anti-VGCC. J Neuroinflammation 2015; 12:167. [PMID: 26377184 PMCID: PMC4574118 DOI: 10.1186/s12974-015-0357-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/02/2015] [Indexed: 01/18/2023] Open
Abstract
Serological testing for anti-neural autoantibodies is important in patients presenting with idiopathic cerebellar ataxia, since these autoantibodies may indicate cancer, determine treatment and predict prognosis. While some of them target nuclear antigens present in all or most CNS neurons (e.g. anti-Hu, anti-Ri), others more specifically target antigens present in the cytoplasm or plasma membrane of Purkinje cells (PC). In this series of articles, we provide a detailed review of the clinical and paraclinical features, oncological, therapeutic and prognostic implications, pathogenetic relevance, and differential laboratory diagnosis of the 12 most common PC autoantibodies (often referred to as 'Medusa head antibodies' due their characteristic somatodendritic binding pattern when tested by immunohistochemistry). To assist immunologists and neurologists in diagnosing these disorders, typical high-resolution immunohistochemical images of all 12 reactivities are presented, diagnostic pitfalls discussed and all currently available assays reviewed. Of note, most of these antibodies target antigens involved in the mGluR1/calcium pathway essential for PC function and survival. Many of the antigens also play a role in spinocerebellar ataxia. Part 1 focuses on anti-metabotropic glutamate receptor 1-, anti-Homer protein homolog 3-, anti-Sj/inositol 1,4,5-trisphosphate receptor- and anti-carbonic anhydrase-related protein VIII-associated autoimmune cerebellar ataxia (ACA); part 2 covers anti-protein kinase C gamma-, anti-glutamate receptor delta-2-, anti-Ca/RhoGTPase-activating protein 26- and anti-voltage-gated calcium channel-associated ACA; and part 3 reviews the current knowledge on anti-Tr/delta notch-like epidermal growth factor-related receptor-, anti-Nb/AP3B2-, anti-Yo/cerebellar degeneration-related protein 2- and Purkinje cell antibody 2-associated ACA, discusses differential diagnostic aspects, and provides a summary and outlook.
Collapse
Affiliation(s)
- S Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, D-69120, Heidelberg, Germany.
| | - B Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, D-69120, Heidelberg, Germany.
| |
Collapse
|
18
|
Lucken-Ardjomande Häsler S, Vallis Y, Jolin HE, McKenzie AN, McMahon HT. GRAF1a is a brain-specific protein that promotes lipid droplet clustering and growth, and is enriched at lipid droplet junctions. J Cell Sci 2014; 127:4602-19. [PMID: 25189622 PMCID: PMC4215711 DOI: 10.1242/jcs.147694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lipid droplets are found in all cell types. Normally present at low levels in the brain, they accumulate in tumours and are associated with neurodegenerative diseases. However, little is known about the mechanisms controlling their homeostasis in the brain. We found that GRAF1a, the longest GRAF1 isoform (GRAF1 is also known as ARHGAP26), was enriched in the brains of neonates. Endogenous GRAF1a was found on lipid droplets in oleic-acid-fed primary glial cells. Exclusive localization required a GRAF1a-specific hydrophobic segment and two membrane-binding regions, a BAR and a PH domain. Overexpression of GRAF1a promoted lipid droplet clustering, inhibited droplet mobility and severely perturbed lipolysis following the chase of cells overloaded with fatty acids. Under these conditions, GRAF1a concentrated at the interface between lipid droplets. Although GRAF1-knockout mice did not show any gross abnormal phenotype, the total lipid droplet volume that accumulated in GRAF1(-/-) primary glia upon incubation with fatty acids was reduced compared to GRAF1(+/+) cells. These results provide additional insights into the mechanisms contributing to lipid droplet growth in non-adipocyte cells, and suggest that proteins with membrane sculpting BAR domains play a role in droplet homeostasis.
Collapse
Affiliation(s)
| | - Yvonne Vallis
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Helen E Jolin
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Andrew N McKenzie
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Harvey T McMahon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
19
|
Cai B, Xie S, Caplan S, Naslavsky N. GRAF1 forms a complex with MICAL-L1 and EHD1 to cooperate in tubular recycling endosome vesiculation. Front Cell Dev Biol 2014; 2:22. [PMID: 25364729 PMCID: PMC4214196 DOI: 10.3389/fcell.2014.00022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 05/08/2014] [Indexed: 12/15/2022] Open
Abstract
The biogenesis of tubular recycling endosomes (TREs) and their subsequent vesiculation after cargo-sorting has occurred, is essential for receptor and lipid recycling to the plasma membrane. Although recent studies have implicated the C-terminal Eps15 Homology Domain (EHD) protein, EHD1, as a key regulator of TRE vesiculation, additional proteins involved in this process have been largely uncharacterized. In the present study, we identify the GTPase Regulator Associated with Focal adhesion kinase-1 (GRAF1) protein in a complex with EHD1 and the TRE hub protein, Molecules Interacting with CasL-Like1 (MICAL-L1). Over-expression of GRAF1 caused vesiculation of MICAL-L1-containing TRE, whereas GRAF1-depletion led to impaired TRE vesiculation and delayed receptor recycling. Moreover, co-addition of purified EHD1 and GRAF1 in a semi-permeabilized cell vesiculation assay produced synergistic TRE vesiculation. Overall, based on our data, we suggest that in addition to its roles in clathrin-independent endocytosis, GRAF1 synergizes with EHD1 to support TRE vesiculation.
Collapse
Affiliation(s)
- Bishuang Cai
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center Omaha, NE, USA
| | - Shuwei Xie
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center Omaha, NE, USA
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center Omaha, NE, USA
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center Omaha, NE, USA
| |
Collapse
|
20
|
Thauerer B, Zur Nedden S, Baier-Bitterlich G. Protein Kinase C-Related Kinase (PKN/PRK). Potential Key-Role for PKN1 in Protection of Hypoxic Neurons. Curr Neuropharmacol 2014; 12:213-8. [PMID: 24851086 PMCID: PMC4023452 DOI: 10.2174/1570159x11666131225000518] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/20/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022] Open
Abstract
Serine/threonine protein kinase C-related kinase (PKN/PRK) is a family of three isoenzymes (PKN1, PKN2,
PKN3), which are widely distributed in eukaryotic organisms and share the same overall domain structure. The Nterminal
region encompasses a conserved repeated domain, termed HR1a-c as well as a HR2/C2 domain. The
serine/threonine kinase domain is found in the C-terminal region of the protein and shows high sequence homology to
other members of the PKC superfamily.
In neurons, PKN1 is the most abundant isoform and has been implicated in a variety of functions including cytoskeletal
organization and neuronal differentiation and its deregulation may contribute to neuropathological processes such as
amyotrophic lateral sclerosis and Alzheimer’s disease. We have recently identified a candidate role of PKN1 in the
regulation of neuroprotective processes during hypoxic stress. Our key findings were that: 1) the activity of PKN1 was
significantly increased by hypoxia (1% O2) and neurotrophins (nerve growth factor and purine nucleosides); 2) Neuronal
cells, deficient of PKN1 showed a decrease of cell viability and neurite formation along with a disturbance of the F-actinassociated
cytoskeleton; 3) Purine nucleoside-mediated neuroprotection during hypoxia was severely hampered in PKN1
deficient neuronal cells, altogether suggesting a potentially critical role of PKN1 in neuroprotective processes.
This review gives an up-to-date overview of the PKN family with a special focus on the neuroprotective role of PKN1 in
hypoxia.
Collapse
Affiliation(s)
- Bettina Thauerer
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Stephanie Zur Nedden
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
21
|
Hutchinson CL, Lowe PN, McLaughlin SH, Mott HR, Owen D. Differential binding of RhoA, RhoB, and RhoC to protein kinase C-related kinase (PRK) isoforms PRK1, PRK2, and PRK3: PRKs have the highest affinity for RhoB. Biochemistry 2013; 52:7999-8011. [PMID: 24128008 DOI: 10.1021/bi401216w] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Protein kinase C-related kinases (PRKs) are members of the protein kinase C superfamily of serine-threonine kinases and can be activated by binding to members of the Rho family of GTPases via a Rho-binding motif known as an HR1 domain. Three tandem HR1 domains reside at the N-terminus of the PRKs. We have assessed the ability of the HR1a and HR1b domains from the three PRK isoforms (PRK1, PRK2, and PRK3) to interact with the three Rho isoforms (RhoA, RhoB, and RhoC). The affinities of RhoA and RhoC for a construct encompassing both PRK1 HR1 domains were similar to those for the HR1a domain alone, suggesting that these interactions are mediated solely by the HR1a domain. The affinities of RhoB for both the PRK1 HR1a domain and the HR1ab didomain were higher than those of RhoA or RhoC. RhoB also bound more tightly to the didomain than to the HR1a domain alone, implicating the HR1b domain in the interaction. As compared with PRK1 HR1 domains, PRK2 and PRK3 domains bind less well to all Rho isoforms. Uniquely, however, the PRK3 domains display a specificity for RhoB that requires both the C-terminus of RhoB and the PRK3 HR1b domain. The thermal stability of the HR1a and HR1b domains was also investigated. The PRK2 HR1a domain was found to be the most thermally stable, while PRK2 HR1b, PRK3 HR1a, and PRK3 HR1b domains all exhibited lower melting temperatures, similar to that of the PRK1 HR1a domain. The lower thermal stability of the PRK2 and PRK3 HR1b domains may impart greater flexibility, driving their ability to interact with Rho isoforms.
Collapse
Affiliation(s)
- Catherine L Hutchinson
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | | | | | | | | |
Collapse
|
22
|
Integrin-mediated signaling induced by simian virus 40 leads to transient uncoupling of cortical actin and the plasma membrane. PLoS One 2013; 8:e55799. [PMID: 23409046 PMCID: PMC3567119 DOI: 10.1371/journal.pone.0055799] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022] Open
Abstract
Simian Virus 40 (SV40) is a paradigm pathogen with multivalent binding sites for the sphingolipid GM1, via which it induces its endocytosis for infection. Here we report that SV40 also utilizes cell surface integrins to activate signaling networks required for infection, even in the absence of the previously implicated glycosphingolipids. We identify ILK, PDK1, the RhoGAP GRAF1 and RhoA as core nodes of the signaling network activated upon SV40 engagement of integrins. We show that integrin-mediated signaling through host SV40 engagement induces the de-phosphorylation of Ezrin leading to uncoupling of the plasma membrane and cortical actin. Our results provide functional evidence for a mechanism by which SV40 activates signal transduction in human epithelial cells via integrins in the context of clathrin-independent endocytosis.
Collapse
|
23
|
Okada H, Uezu A, Mason FM, Soderblom EJ, Moseley MA, Soderling SH. SH3 domain-based phototrapping in living cells reveals Rho family GAP signaling complexes. Sci Signal 2011; 4:rs13. [PMID: 22126966 DOI: 10.1126/scisignal.2002189] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Rho family GAPs [guanosine triphosphatase (GTPase) activating proteins] negatively regulate Rho family GTPase activity and therefore modulate signaling events that control cytoskeletal dynamics. The spatial distribution of these GAPs and their specificity toward individual GTPases are controlled by their interactions with various proteins within signaling complexes. These interactions are likely mediated through the Src homology 3 (SH3) domain, which is abundant in the Rho family GAP proteome and exhibits a micromolar binding affinity, enabling the Rho family GAPs to participate in transient interactions with multiple binding partners. To capture these elusive GAP signaling complexes in situ, we developed a domain-based proteomics approach, starting with in vivo phototrapping of SH3 domain-binding proteins and the mass spectrometry identification of associated proteins for nine representative Rho family GAPs. After the selection of candidate binding proteins by cluster analysis, we performed peptide array-based high-throughput in vitro binding assays to confirm the direct interactions and map the SH3 domain-binding sequences. We thereby identified 54 SH3-mediated binding interactions (including 51 previously unidentified ones) for nine Rho family GAPs. We constructed Rho family GAP interactomes that provided insight into the functions of these GAPs. We further characterized one of the predicted functions for the Rac-specific GAP WRP and identified a role for WRP in mediating clustering of the postsynaptic scaffolding protein gephyrin and the GABA(A) (γ-aminobutyric acid type A) receptor at inhibitory synapses.
Collapse
Affiliation(s)
- Hirokazu Okada
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
24
|
Lachmann S, Jevons A, De Rycker M, Casamassima A, Radtke S, Collazos A, Parker PJ. Regulatory domain selectivity in the cell-type specific PKN-dependence of cell migration. PLoS One 2011; 6:e21732. [PMID: 21754995 PMCID: PMC3130767 DOI: 10.1371/journal.pone.0021732] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 06/09/2011] [Indexed: 12/30/2022] Open
Abstract
The mammalian protein kinase N (PKN) family of Serine/Threonine kinases comprises three isoforms, which are targets for Rho family GTPases. Small GTPases are major regulators of the cellular cytoskeleton, generating interest in the role(s) of specific PKN isoforms in processes such as cell migration and invasion. It has been reported that PKN3 is required for prostate tumour cell invasion but not PKN1 or 2. Here we employ a cell model, the 5637 bladder tumour cell line where PKN2 is relatively highly expressed, to assess the potential redundancy of these isoforms in migratory responses. It is established that PKN2 has a critical role in the migration and invasion of these cells. Furthermore, using a PKN wild-type and chimera rescue strategy, it is shown that PKN isoforms are not simply redundant in supporting migration, but appear to be linked through isoform specific regulatory domain properties to selective upstream signals. It is concluded that intervention in PKNs may need to be directed at multiple isoforms to be effective in different cell types.
Collapse
Affiliation(s)
- Sylvie Lachmann
- Protein Phosphorylation Laboratory, London Research Institute, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
25
|
Barrios J, Wieder R. Dual FGF-2 and intergrin alpha5beta1 signaling mediate GRAF-induced RhoA inactivation in a model of breast cancer dormancy. CANCER MICROENVIRONMENT 2009; 2:33-47. [PMID: 19308677 PMCID: PMC2787927 DOI: 10.1007/s12307-009-0019-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 01/14/2009] [Indexed: 12/25/2022]
Abstract
Interactions with the bone marrow stroma regulate dormancy and survival of breast cancer micrometastases. In an in vitro model of dormancy in the bone marrow, we previously demonstrated that estrogen-dependent breast cancer cells are partially re-differentiated by FGF-2, re-express integrin α5β1 lost with malignant transformation and acquire an activated PI3K/Akt pathway. Ligation of integrin α5β1 by fibronectin and activation of the PI3K pathway both contribute to survival of these dormant cells. Here, we investigated mechanisms responsible for the dormant phenotype. Experiments demonstrate that integrin α5β1 controls de novo cytoskeletal rearrangements, cell spreading, focal adhesion kinase rearrangement to the cell perimeter and recruitment of a RhoA GAP known as GRAF. This results in the inactivation of RhoA, an effect which is necessary for the stabilization of cortical actin. Experiments also demonstrate that activation of the PI3K pathway by FGF-2 is independent of integrin α5β1 and is also required for cortical actin reorganization, GRAF membrane relocalization and RhoA inactivation. These data suggest that GRAF-mediated RhoA inactivation and consequent phenotypic changes of dormancy depend on dual signaling by FGF-2-initiated PI3K activation and through ligation of integrin α5β1 by fibronectin.
Collapse
Affiliation(s)
- Judith Barrios
- Department of Medicine, UMDNJ-New Jersey Medical School-University Hospital Cancer Center, 185 South Orange Avenue, CCH1216, Newark, NJ 07103 USA
| | - Robert Wieder
- Department of Medicine, UMDNJ-New Jersey Medical School-University Hospital Cancer Center, 185 South Orange Avenue, CCH1216, Newark, NJ 07103 USA
| |
Collapse
|
26
|
Lundmark R, Doherty GJ, Howes MT, Cortese K, Vallis Y, Parton RG, McMahon HT. The GTPase-activating protein GRAF1 regulates the CLIC/GEEC endocytic pathway. Curr Biol 2009; 18:1802-8. [PMID: 19036340 PMCID: PMC2726289 DOI: 10.1016/j.cub.2008.10.044] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 10/04/2008] [Accepted: 10/08/2008] [Indexed: 10/29/2022]
Abstract
Clathrin-independent endocytosis is an umbrella term for a variety of endocytic pathways that internalize numerous cargoes independently of the canonical coat protein Clathrin [1, 2]. Electron-microscopy studies have defined the pleiomorphic CLathrin-Independent Carriers (CLICs) and GPI-Enriched Endocytic Compartments (GEECs) as related major players in such uptake [3, 4]. This CLIC/GEEC pathway relies upon cellular signaling and activation through small G proteins, but mechanistic insight into the biogenesis of its tubular and tubulovesicular carriers is lacking. Here we show that the Rho-GAP-domain-containing protein GRAF1 marks, and is indispensable for, a major Clathrin-independent endocytic pathway. This pathway is characterized by its ability to internalize bacterial exotoxins, GPI-linked proteins, and extracellular fluid. We show that GRAF1 localizes to PtdIns(4,5)P2-enriched, tubular, and punctate lipid structures via N-terminal BAR and PH domains. These membrane carriers are relatively devoid of caveolin1 and flotillin1 but are associated with activity of the small G protein Cdc42. This study provides the first specific noncargo marker for CLIC/GEEC endocytic membranes and demonstrates how GRAF1 can coordinate small G protein signaling and membrane remodeling to facilitate internalization of CLIC/GEEC pathway cargoes.
Collapse
Affiliation(s)
- Richard Lundmark
- Medical Research Council, Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 0QH, UK.
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The Rho GTPases are implicated in almost every fundamental cellular process. They act as molecular switches that cycle between an active GTP-bound and an inactive GDP-bound state. Their slow intrinsic GTPase activity is greatly enhanced by RhoGAPs (Rho GTPase-activating proteins), thus causing their inactivation. To date, more than 70 RhoGAPs have been identified in eukaryotes, ranging from yeast to human, and based on sequence homology of their RhoGAP domain, we have grouped them into subfamilies. In the present Review, we discuss their regulation, biological functions and implication in human diseases.
Collapse
Affiliation(s)
- Joseph Tcherkezian
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 2B2
| | | |
Collapse
|
28
|
Betson M, Settleman J. A rho-binding protein kinase C-like activity is required for the function of protein kinase N in Drosophila development. Genetics 2007; 176:2201-12. [PMID: 17507675 PMCID: PMC1950625 DOI: 10.1534/genetics.107.072967] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Rho GTPases interact with multiple downstream effectors to exert their biological functions, which include important roles in tissue morphogenesis during the development of multicellular organisms. Among the Rho effectors are the protein kinase N (PKN) proteins, which are protein kinase C (PKC)-like kinases that bind activated Rho GTPases. The PKN proteins are well conserved evolutionarily, but their biological role in any organism is poorly understood. We previously determined that the single Drosophila ortholog of mammalian PKN proteins, Pkn, is a Rho/Rac-binding kinase essential for Drosophila development. By performing "rescue" studies with various Pkn mutant constructs, we have defined the domains of Pkn required for its role during Drosophila development. These studies suggested that Rho, but not Rac binding is important for Pkn function in development. In addition, we determined that the kinase domain of PKC53E, a PKC family kinase, can functionally substitute for the kinase domain of Pkn during development, thereby exemplifying the evolutionary strategy of "combining" functional domains to produce proteins with distinct biological activities. Interestingly, we also identified a requirement for Pkn in wing morphogenesis, thereby revealing the first postembryonic function for Pkn.
Collapse
Affiliation(s)
- Martha Betson
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA 02129, USA
| | | |
Collapse
|
29
|
Abstract
Among the key protein regulators of the various and numerous small GTPases are the GTPase activating proteins (GAPs). Experimental studies of some of the approximately 170 GAPs predicted by the human genome indicate that their catalytic GAP activity is regulated by a variety of mechanisms, including phosphorylation, protein-protein interactions, proteolysis, and interactions with lipids. Most reported biochemical studies to address the specificity of GAPs for particular GTPases have been conducted in vitro with bacterially produced GTPases. Thus, the potential influence of these various regulatory mechanisms in the context of GAP-GTPase specificity may be overlooked in such assays. Here, we present experimental studies that highlight the role of lipids in modulating the GTPase specificity for some of the Rho GAPs. We find that particular phospholipids can substantially alter the substrate "preference" for the p190 GAPs. We find that C-terminal prenylation of GTPases can influence the specificity of GAP interactions as well. These observations emphasize the limitations of standard in vitro GAP assays in definitively establishing the physiologically relevant GTPase targets for particular GAPs.
Collapse
Affiliation(s)
- Erzsébet Ligeti
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | | |
Collapse
|
30
|
Zhao ZS, Manser E. PAK and other Rho-associated kinases--effectors with surprisingly diverse mechanisms of regulation. Biochem J 2005; 386:201-14. [PMID: 15548136 PMCID: PMC1134783 DOI: 10.1042/bj20041638] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 11/09/2004] [Accepted: 11/17/2004] [Indexed: 11/17/2022]
Abstract
The Rho GTPases are a family of molecular switches that are critical regulators of signal transduction pathways in eukaryotic cells. They are known principally for their role in regulating the cytoskeleton, and do so by recruiting a variety of downstream effector proteins. Kinases form an important class of Rho effector, and part of the biological complexity brought about by switching on a single GTPase results from downstream phosphorylation cascades. Here we focus on our current understanding of the way in which different Rho-associated serine/threonine kinases, denoted PAK (p21-activated kinase), MLK (mixed-lineage kinase), ROK (Rho-kinase), MRCK (myotonin-related Cdc42-binding kinase), CRIK (citron kinase) and PKN (protein kinase novel), interact with and are regulated by their partner GTPases. All of these kinases have in common an ability to dimerize, and in most cases interact with a variety of other proteins that are important for their function. A diversity of known structures underpin the Rho GTPase-kinase interaction, but only in the case of PAK do we have a good molecular understanding of kinase regulation. The ability of Rho GTPases to co-ordinate spatial and temporal phosphorylation events explains in part their prominent role in eukaryotic cell biology.
Collapse
Key Words
- cdc42
- mlk (mixed-lineage kinase)
- pak (p21-activated kinase)
- rac
- rho
- rok (rho-kinase)
- acc, anti-parallel coiled-coil
- crib, cdc42 and rac interactive binding
- crik, citron kinase
- crmp, collapsin response mediator protein
- dmpk, myotonic dystrophy kinase
- gef, guanine nucleotide exchange factor
- git1, g-protein-coupled receptor kinase-interacting target 1
- hsp90, heat shock protein 90
- jnk, c-jun n-terminal kinase
- ki, kinase inhibitory
- kim, ki motif
- limk, lim domain kinase
- mapk, mitogen-activated protein kinase
- mbs, myosin-binding subunit
- mekk, mapk/erk (extracellular-signal-regulated kinase) kinase kinase
- mkk, mapk kinase
- mlk, mixed-lineage kinase
- mrck, myotonin-related cdc42-binding kinase
- pak, p21-activated kinase
- pbd, p21-binding domain
- pdk1, 3-phosphoinositide-dependent kinase 1
- ph, pleckstrin homology
- pix, pak-interacting exchange factor
- pkc, protein kinase c
- pkn, protein kinase novel
- pp1, protein phosphatase type 1
- r-mlc, regulatory myosin light chain
- rok, rho-kinase
- sh3, src homology 3
Collapse
Affiliation(s)
- Zhou-shen Zhao
- GSK-IMCB Laboratory, Institute of Molecular and Cell Biology, Proteos Building, 61 Biopolis Drive, Singapore 138673
| | - Ed Manser
- GSK-IMCB Laboratory, Institute of Molecular and Cell Biology, Proteos Building, 61 Biopolis Drive, Singapore 138673
| |
Collapse
|
31
|
Lavelin I, Geiger B. Characterization of a novel GTPase-activating protein associated with focal adhesions and the actin cytoskeleton. J Biol Chem 2004; 280:7178-85. [PMID: 15611138 DOI: 10.1074/jbc.m411990200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the present study we characterize a novel RhoGAP protein (RC-GAP72) that interacts with actin stress fibers, focal adhesions, and cell-cell adherens junctions via its 185-amino acid C-terminal region. Overexpression of RC-GAP72 in fibroblasts induces cell rounding with partial or complete disruption of actin stress fibers and formation of membrane ruffles, lamellipodia, and filopodia. RC-GAP72 mutant truncated downstream of the GTPase-activating protein (GAP) domain retains the ability to stimulate membrane protrusions but fails to affect stress fiber integrity or induce cell retraction. A mutant protein consisting of the C terminus of RC-GAP72 and lacking the GAP domain does not exert any visible effect on cellular morphology. Inactivation of the GAP domain by a point mutation does not abolish the effect of RC-GAP72 on actin stress fibers but moderates its capability to induce membrane protrusions. Our data imply that the cytoskeletal localization of RC-GAP72 and its interaction with GTPases are essential for its effect on the integrity of actin stress fibers, whereas the induction of lamellipodia and filopodia depends on the activity of the GAP domain irrespective of binding to the actin cytoskeleton. We propose that RC-GAP72 affects cellular morphology by targeting activated Cdc42 and Rac1 GTPases to specific subcellular sites, triggering local morphological changes. The overall physiological functions of RC-GAP72 are presently unknown, yet our data suggest that RC-GAP72 plays a role in regulating cell morphology and cytoskeletal organization.
Collapse
Affiliation(s)
- Irena Lavelin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
32
|
Gotoh Y, Oishi K, Shibata H, Yamagiwa A, Isagawa T, Nishimura T, Goyama E, Takahashi M, Mukai H, Ono Y. Protein kinase PKN1 associates with TRAF2 and is involved in TRAF2-NF-κB signaling pathway. Biochem Biophys Res Commun 2004; 314:688-94. [PMID: 14741690 DOI: 10.1016/j.bbrc.2003.12.148] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
PKN1 is a fatty acid and Rho-activated serine/threonine protein kinase whose catalytic domain is highly homologous to protein kinase C (PKC) family. In yeast two-hybrid screening for PKN1 binding proteins, we identified tumor necrosis factor alpha (TNFalpha) receptor-associated factor 2 (TRAF2). TRAF2 is one of the major mediators of TNF receptor superfamily transducing TNF signal to various functional targets, including activation of NF-kappaB, JNK, and apoptosis. FLAG-tagged PKN1 was co-immunoprecipitated with endogenous TRAF2 from HEK293 cell lysate, and in vitro binding assay using the deletion mutants of TRAF2 showed that PKN1 directly binds to the TRAF domain of TRAF2. PKN1 has the TRAF2-binding consensus sequences PXQX (S/T) at amino acid residues 580-584 (PIQES), and P580AQ582A mutant was not co-immunoprecipitated with TRAF2. Furthermore, the reduced expression of PKN1 by RNA interference (RNAi) down-regulated TRAF2-induced NF-kappaB activation in HEK293T cells. These results suggest that PKN1 is involved in TRAF2-NF-kappaB signaling pathway.
Collapse
Affiliation(s)
- Yusuke Gotoh
- Graduate School of Science and Technology, Kobe University, 657-8501, Kobe, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bernards A. GAPs galore! A survey of putative Ras superfamily GTPase activating proteins in man and Drosophila. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1603:47-82. [PMID: 12618308 DOI: 10.1016/s0304-419x(02)00082-3] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Typical members of the Ras superfamily of small monomeric GTP-binding proteins function as regulators of diverse processes by cycling between biologically active GTP- and inactive GDP-bound conformations. Proteins that control this cycling include guanine nucleotide exchange factors or GEFs, which activate Ras superfamily members by catalyzing GTP for GDP exchange, and GTPase activating proteins or GAPs, which accelerate the low intrinsic GTP hydrolysis rate of typical Ras superfamily members, thus causing their inactivation. Two among the latter class of proteins have been implicated in common genetic disorders associated with an increased cancer risk, neurofibromatosis-1, and tuberous sclerosis. To facilitate genetic analysis, I surveyed Drosophila and human sequence databases for genes predicting proteins related to GAPs for Ras superfamily members. Remarkably, close to 0.5% of genes in both species (173 human and 64 Drosophila genes) predict proteins related to GAPs for Arf, Rab, Ran, Rap, Ras, Rho, and Sar family GTPases. Information on these genes has been entered into a pair of relational databases, which can be used to identify evolutionary conserved proteins that are likely to serve basic biological functions, and which can be updated when definitive information on the coding potential of both genomes becomes available.
Collapse
Affiliation(s)
- André Bernards
- Massachusetts General Hospital Cancer Center, Building 149, 13th Street, Charlestown, MA 02129-2000, USA.
| |
Collapse
|
34
|
Peck J, Douglas G, Wu CH, Burbelo PD. Human RhoGAP domain-containing proteins: structure, function and evolutionary relationships. FEBS Lett 2002; 528:27-34. [PMID: 12297274 DOI: 10.1016/s0014-5793(02)03331-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Proteins containing a RhoGAP (Rho GTPase activating protein) domain usually function to catalyze the hydrolysis of GTP that is bound to Rho, Rac and/or Cdc42, inactivating these regulators of the actin cytoskeleton. Using database searches, at least 53 distinct RhoGAP domain-containing proteins are likely to be encoded in human DNA. Phylogenetic analysis of only the RhoGAP domains divides these proteins into distinct families that appear to be functionally related. We also review the current understanding of the structure and likely functions of these human proteins. The presence of RhoGAP domains in a number of different human proteins suggests that cytoskeletal changes, regulated by Rho GTPase, may be integrated with many different signaling pathways.
Collapse
Affiliation(s)
- Jeremy Peck
- Department of Oncology, Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
35
|
Kitaura Y, Satoh H, Takahashi H, Shibata H, Maki M. Both ALG-2 and peflin, penta-EF-hand (PEF) proteins, are stabilized by dimerization through their fifth EF-hand regions. Arch Biochem Biophys 2002; 399:12-8. [PMID: 11883899 DOI: 10.1006/abbi.2001.2736] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
ALG-2 (apoptosis-linked gene-2 protein) and peflin are Ca(2+)-binding proteins and belong to the penta-EF-hand (PEF) protein family, which includes calpain, sorcin, and grancalcin. ALG-2 forms either a homodimer or a heterodimer with peflin like other PEF proteins. In this study, we found that the fifth-EF-hand (EF-5) regions of both ALG-2 and peflin are essential for dimerization and their stabilities. Exogenously expressed EF-5-deletion (DeltaEF-5) mutants of ALG-2 and peflin were unstable and were not detected in HEK293 cells by Western blotting. In a pulse--chase experiment, the DeltaEF-5 mutants were rapidly degraded, but they were stabilized by treatment with a proteasome inhibitor, MG132. In MG132-treated cells, DeltaEF-5 mutants were recovered in the insoluble fractions. Transient coexpression of ALG-2 increased the peflin level. These results indicate that the absence of a fifth EF-hand results in rapid degradation by the proteasome. On the other hand, stable expression of exogenous peflin decreased the amount of endogenous peflin. The amount of peflin that can dimerize with ALG-2 seems to be restricted in mammalian cells.
Collapse
Affiliation(s)
- Yasuyuki Kitaura
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | | | | | | | | |
Collapse
|
36
|
Aoto H, Sasaki H, Ishino M, Sasaki T. Nuclear translocation of cell adhesion kinase beta/proline-rich tyrosine kinase 2. Cell Struct Funct 2002; 27:47-61. [PMID: 11937718 DOI: 10.1247/csf.27.47] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cell adhesion kinase beta (CAKbeta/PYK2) is a protein-tyrosine kinase of the focal adhesion kinase (FAK) family. Whereas FAK predominantly localizes at focal adhesions, CAK beta localizes at the perinuclear region in fibroblasts. Here we expressed in cultured cells two point mutants of CAKbeta, P717A and P859A, each of which had lost one of its two PXXP motifs, the ligand sequence for SH3 domains, found at the CAKbeta C-terminal region. We observed a remarkable change in the subcellular distribution of the P859A mutant; while that of the P717A mutant was the same as the wild type. The P859A mutant localized exclusively in the cell nucleus in all cell lines examined. Wild-type CAKbeta also accumulated in the nucleus when cells were treated with an inhibitor of the nuclear export of proteins. These results indicate that CAK beta shuttles between the cytoplasm and the nucleus. On nuclear accumulation of P859A-CAKbeta, a CAKbeta-binding protein, Hic-5, also accumulated in the nucleus. P859A-CAKbeta and co-expressed Hic-5 formed nuclear speckles, in which one other CAK beta-binding protein, p130(Cas), was also concentrated. These findings on nuclear translocation of CAK beta imply that CAKbeta may regulate nuclear processes such as transcription, particularly because Hic-5 was recently shown to be a coactivator of nuclear receptors.
Collapse
Affiliation(s)
- Hiroshi Aoto
- Department of Biochemistry, Cancer Research Institute, Sapporo Medical University, Japan.
| | | | | | | |
Collapse
|